Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Pharmacol Exp Ther ; 387(2): 226-234, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37679045

RESUMEN

The type-5 muscarinic acetylcholine receptor (mAChR, M5) is almost exclusively expressed in dopamine (DA) neurons of the ventral tegmental area and substantia nigra pars compacta; therefore, they are ideally located to modulate DA signaling and underlying behaviors. However, the role of M5 in shaping DA release is still poorly characterized. In this study, we first quantitatively mapped the expression of M5 in different neurons of the mouse midbrain, then used voltammetry in mouse striatum to evaluate the effect of M5-selective modulators on DA release. The M5 negative allosteric modulator ML375 significantly decreased electrically evoked DA release and blocked the effect of Oxotremorine-M (Oxo-M; nonselective mAChR agonist) on DA release in the presence of an acetylcholine nicotinic receptor blocker. Conversely, the M5 positive allosteric modulator VU 0365114 significantly increased electrically evoked DA release and the Oxo-M effect on DA release. We then assessed M5's impact on mesolimbic circuit function in vivo. Although psychostimulant-induced locomotor activity models in knockout mice have previously been used to characterize the role of M5 in DA transmission, the results of these studies conflict, leading us to select a different in vivo model, namely a cocaine self-administration paradigm. In contrast to a previous study that also used this model, in the current study, administration of ML375 did not decrease cocaine self-administration in rats (using fixed and progressive ratio). These conflicting results illustrate the complexity of M5 modulation and the need to further characterize its involvement in the regulation of dopamine signaling, central to multiple neuropsychiatric diseases. SIGNIFICANCE STATEMENT: This work describes the type-5 muscarinic receptor (M5) pattern of expression within the midbrain as well as its physiological modulation by selective compounds at the axon terminal level in the striatum, where M5 directly shapes dopamine transmission. It offers the first direct readout of mesolimbic dopamine release modulation by M5, highlighting its role in regulating neurocircuits implicated in the pathophysiology of neuropsychiatric disorders such as substance use disorders, major depressive disorder, and schizophrenia.

2.
PLoS One ; 15(10): e0240991, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33091047

RESUMEN

Human induced Pluripotent Stem Cells (iPSCs) are a powerful tool to dissect the biology of complex human cell types such as those of the central nervous system (CNS). However, robust, high-throughput platforms for reliably measuring activity in human iPSC-derived neuronal cultures are lacking. Here, we assessed 3D cultures of cortical neurons and astrocytes displaying spontaneous, rhythmic, and highly synchronized neural activity that can be visualized as calcium oscillations on standard high-throughput fluorescent readers as a platform for CNS-based discovery efforts. Spontaneous activity and spheroid structure were highly consistent from well-to-well, reference compounds such as TTX, 4-AP, AP5, and NBQX, had expected effects on neural spontaneous activity, demonstrating the presence of functionally integrated neuronal circuitry. Neurospheroid biology was challenged by screening the LOPAC®1280 library, a collection of 1280 pharmacologically active small molecules. The primary screen identified 111 compounds (8.7%) that modulated neural network activity across a wide range of neural and cellular processes and 16 of 17 compounds chosen for follow-up confirmed the primary screen results. Together, these data demonstrate the suitability and utility of human iPSC-derived neurospheroids as a screening platform for CNS-based drug discovery.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Astrocitos/citología , Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/citología , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Tamizaje Masivo/métodos , Células-Madre Neurales/citología
3.
Nat Genet ; 51(12): 1691-1701, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31740836

RESUMEN

In the mammalian genome, the clustered protocadherin (cPCDH) locus provides a paradigm for stochastic gene expression with the potential to generate a unique cPCDH combination in every neuron. Here we report a chromatin-based mechanism that emerges during the transition from the naive to the primed states of cell pluripotency and reduces, by orders of magnitude, the combinatorial potential in the human cPCDH locus. This mechanism selectively increases the frequency of stochastic selection of a small subset of cPCDH genes after neuronal differentiation in monolayers, 10-month-old cortical organoids and engrafted cells in the spinal cords of rats. Signs of these frequent selections can be observed in the brain throughout fetal development and disappear after birth, except in conditions of delayed maturation such as Down's syndrome. We therefore propose that a pattern of limited cPCDH-gene expression diversity is maintained while human neurons still retain fetal-like levels of maturation.


Asunto(s)
Cadherinas/genética , Cromatina/genética , Síndrome de Down/patología , Células Madre Pluripotentes Inducidas/citología , Neuronas/fisiología , Adulto , Animales , Astrocitos/citología , Astrocitos/fisiología , Encéfalo/citología , Encéfalo/embriología , Diferenciación Celular , Línea Celular , Síndrome de Down/genética , Regulación de la Expresión Génica , Histonas/genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Persona de Mediana Edad , Neuronas/citología , Regiones Promotoras Genéticas , Ratas , Análisis de la Célula Individual , Médula Espinal/citología , Médula Espinal/trasplante , Trasplante Heterólogo
4.
Mol Pharmacol ; 94(3): 1092-1100, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29980658

RESUMEN

Zinc homeostasis is a highly regulated process in mammalian cells that is critical for normal growth and development. Movement of zinc across cell compartments is controlled by two classes of transporters: Slc39a family members transport zinc into the cytosol from either the extracellular space or intracellular stores such as the endoplasmic reticulum (ER), whereas the SLC30A family mediates zinc efflux from the cytosol. In this study, we report that genetic ablation of SLC39A7 (ZIP7) results in decreased cytosolic zinc levels, increased ER zinc levels, impaired cell proliferation, and induction of ER stress. Confirmatory of impaired zinc transport as the causal mechanism, both the increased ER stress and impaired cell proliferation were rescued by increasing cytosolic zinc. Furthermore, using these robust cellular phenotypes, we implemented a small-molecule library screen with 2800 compounds and identified one small molecule capable of rescuing ER stress and cell proliferation in ZIP7-deficient cells in the low micromolar range.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Zinc/metabolismo , Proteínas de Transporte de Catión/genética , Línea Celular , Proliferación Celular/fisiología , Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Humanos
5.
J Biol Chem ; 293(29): 11341-11357, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-29858247

RESUMEN

Mounting evidence suggests that alterations in cholesterol homeostasis are involved in Alzheimer's disease (AD) pathogenesis. Amyloid precursor protein (APP) or multiple fragments generated by proteolytic processing of APP have previously been implicated in the regulation of cholesterol metabolism. However, the physiological function of APP in regulating lipoprotein homeostasis in astrocytes, which are responsible for de novo cholesterol biosynthesis and regulation in the brain, remains unclear. To address this, here we used CRISPR/Cas9 genome editing to generate isogenic APP-knockout (KO) human induced pluripotent stem cells (hiPSCs) and differentiated them into human astrocytes. We found that APP-KO astrocytes have reduced cholesterol and elevated levels of sterol regulatory element-binding protein (SREBP) target gene transcripts and proteins, which were both downstream consequences of reduced lipoprotein endocytosis. To elucidate which APP fragments regulate cholesterol homeostasis and to examine whether familial AD mutations in APP affect lipoprotein metabolism, we analyzed an isogenic allelic series harboring the APP Swedish and APP V717F variants. Only astrocytes homozygous for the APP Swedish (APPSwe/Swe) mutation, which had reduced full-length APP (FL APP) due to increased ß-secretase cleavage, recapitulated the APP-KO phenotypes. Astrocytic internalization of ß-amyloid (Aß), another ligand for low-density lipoprotein (LDL) receptors, was also impaired in APP-KO and APPSwe/Swe astrocytes. Finally, impairing cleavage of FL APP through ß-secretase inhibition in APPSwe/Swe astrocytes reversed the LDL and Aß endocytosis defects. In conclusion, FL APP is involved in the endocytosis of LDL receptor ligands and is required for proper cholesterol homeostasis and Aß clearance in human astrocytes.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Astrocitos/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Precursor de Proteína beta-Amiloide/genética , Astrocitos/citología , Sistemas CRISPR-Cas , Línea Celular , Endocitosis , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Receptores de LDL/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
6.
Sci Transl Med ; 10(442)2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29794059

RESUMEN

Axon regeneration after spinal cord injury (SCI) is attenuated by growth inhibitory molecules associated with myelin. We report that rat myelin stimulated the growth of axons emerging from rat neural progenitor cells (NPCs) transplanted into sites of SCI in adult rat recipients. When plated on a myelin substrate, neurite outgrowth from rat NPCs and from human induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) was enhanced threefold. In vivo, rat NPCs and human iPSC-derived NSCs extended greater numbers of axons through adult central nervous system white matter than through gray matter and preferentially associated with rat host myelin. Mechanistic investigations excluded Nogo receptor signaling as a mediator of stem cell-derived axon growth in response to myelin. Transcriptomic screens of rodent NPCs identified the cell adhesion molecule neuronal growth regulator 1 (Negr1) as one mediator of permissive axon-myelin interactions. The stimulatory effect of myelin-associated proteins on rodent NPCs was developmentally regulated and involved direct activation of the extracellular signal-regulated kinase (ERK). The stimulatory effects of myelin on NPC/NSC axon outgrowth should be investigated further and could potentially be exploited for neural repair after SCI.


Asunto(s)
Envejecimiento/metabolismo , Axones/metabolismo , Vaina de Mielina/metabolismo , Células-Madre Neurales/citología , Proyección Neuronal , Animales , Axones/ultraestructura , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Sustancia Gris/citología , Humanos , Ratones Endogámicos C57BL , Vaina de Mielina/ultraestructura , Células-Madre Neurales/ultraestructura , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Endogámicas F344 , Ratas Desnudas , Médula Espinal/citología , Sustancia Blanca/citología
7.
Stem Cell Reports ; 8(4): 1101-1111, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28410643

RESUMEN

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) offers the possibility of studying the molecular mechanisms underlying human diseases in cell types difficult to extract from living patients, such as neurons and cardiomyocytes. To date, studies have been published that use small panels of iPSC-derived cell lines to study monogenic diseases. However, to study complex diseases, where the genetic variation underlying the disorder is unknown, a sizable number of patient-specific iPSC lines and controls need to be generated. Currently the methods for deriving and characterizing iPSCs are time consuming, expensive, and, in some cases, descriptive but not quantitative. Here we set out to develop a set of simple methods that reduce cost and increase throughput in the characterization of iPSC lines. Specifically, we outline methods for high-throughput quantification of surface markers, gene expression analysis of in vitro differentiation potential, and evaluation of karyotype with markedly reduced cost.


Asunto(s)
Variación Genética , Ensayos Analíticos de Alto Rendimiento/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación/métodos , Miocitos Cardíacos/metabolismo , Neuronas/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Reprogramación Celular/genética , Análisis Costo-Beneficio , Genotipo , Ensayos Analíticos de Alto Rendimiento/economía , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Células Madre Pluripotentes Inducidas/citología , Cariotipificación/economía , Miocitos Cardíacos/citología , Neuronas/citología , Fenotipo
8.
Pharmacol Res ; 115: 233-241, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27832960

RESUMEN

Indole-based compounds, such as the alkyl-indole (AI) compound WIN55212-2, activate the cannabinoid receptors, CB1 and CB2, two well-characterized G protein-coupled receptors (GPCR). Reports indicate that several indole-based cannabinoid agonists, including WIN55212-2, lack selectivity and interact with at least two additional targets: AI-sensitive GPCRs and microtubules. Studying how indole-based compounds modulate the activity of these 4 targets has been difficult as selective chemical tools were not available. Here we report the pharmacological characterization of six newly-developed indole-based compounds (ST-11, ST-23, ST-25, ST-29, ST-47 and ST-48) that exhibit distinct binding affinities at AI-sensitive receptors, cannabinoid CB1 and CB2 receptors and the colchicine site of tubulin. Several compounds exhibit some level of selectivity for AI-sensitive receptors, including ST-11 that binds AI-sensitive receptors with a Kd of 52nM and appears to have a weaker affinity for the colchicine site of tubulin (Kd=3.2µM) and does not bind CB1/CB2 receptors. Leveraging these characteristics, we show that activation of AI-sensitive receptors with ST-11 inhibits both the basal and stimulated migration of the Delayed Brain Tumor (DBT) mouse glioma cell line. Our study describes a new series of indole-based compounds that enable the pharmacological and functional differentiation of alkylindole-sensitive receptors from cannabinoid receptors and microtubules.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Glioma/tratamiento farmacológico , Indoles/farmacología , Microtúbulos/efectos de los fármacos , Receptores de Cannabinoides/metabolismo , Animales , Benzoxazinas/farmacología , Unión Competitiva/fisiología , Agonistas de Receptores de Cannabinoides/farmacología , Línea Celular , Colchicina/metabolismo , Glioma/metabolismo , Células HEK293 , Humanos , Ratones , Morfolinas/farmacología , Naftalenos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Tubulina (Proteína)/metabolismo
9.
Cell Rep ; 17(3): 759-773, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27732852

RESUMEN

We investigated early phenotypes caused by familial Alzheimer's disease (fAD) mutations in isogenic human iPSC-derived neurons. Analysis of neurons carrying fAD PS1 or APP mutations introduced using genome editing technology at the endogenous loci revealed that fAD mutant neurons had previously unreported defects in the recycling state of endocytosis and soma-to-axon transcytosis of APP and lipoproteins. The endocytosis reduction could be rescued through treatment with a ß-secretase inhibitor. Our data suggest that accumulation of ß-CTFs of APP, but not Aß, slow vesicle formation from an endocytic recycling compartment marked by the transcytotic GTPase Rab11. We confirm previous results that endocytosis is affected in AD and extend these to uncover a neuron-specific defect. Decreased lipoprotein endocytosis and transcytosis to the axon suggest that a neuron-specific impairment in endocytic axonal delivery of lipoproteins and other key materials might compromise synaptic maintenance in fAD.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Lipoproteínas LDL/metabolismo , Mutación/genética , Neuronas/metabolismo , Transcitosis , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Axones/metabolismo , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Presenilina-1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al GTP rab/metabolismo
10.
Cell Stem Cell ; 16(4): 373-85, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25772071

RESUMEN

Predisposition to sporadic Alzheimer's disease (SAD) involves interactions between a person's unique combination of genetic variants and the environment. The molecular effect of these variants may be subtle and difficult to analyze with standard in vitro or in vivo models. Here we used hIPSCs to examine genetic variation in the SORL1 gene and possible contributions to SAD-related phenotypes in human neurons. We found that human neurons carrying SORL1 variants associated with an increased SAD risk show a reduced response to treatment with BDNF, at the level of both SORL1 expression and APP processing. shRNA knockdown of SORL1 demonstrates that the differences in BDNF-induced APP processing between genotypes are dependent on SORL1 expression. We propose that the variation in SORL1 expression induction by BDNF is modulated by common genetic variants and can explain how genetic variation in this one locus can contribute to an individual's risk of developing SAD.


Asunto(s)
Enfermedad de Alzheimer/genética , Células Madre Pluripotentes Inducidas/fisiología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas de Transporte de Membrana/genética , Neuronas/fisiología , Proteína Amiloide A Sérica/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Análisis Mutacional de ADN/métodos , Regulación de la Expresión Génica/genética , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Fenotipo , Polimorfismo Genético , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , Factores de Riesgo
11.
Neurotherapeutics ; 12(1): 121-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25534395

RESUMEN

Our understanding of Alzheimer's disease (AD) is still incomplete and, as a result, we lack effective therapies. Reprogramming to generate human-induced pluripotent stem cells provides a new approach to the generation of human neurons that carry the genomes of people with familial or sporadic AD. Differentiation of such stem cells to human neurons is already providing new insights into AD and molecular pathways that may provide new targets for effective therapy. These pathways include typical amyloid response pathways, as well as pathways leading from altered behavior of amyloid precursor protein to the elevated phosphorylation of tau protein. There is also a need for standardization of models so that isogenic lines differing only in the familial AD mutation can be compared.


Asunto(s)
Enfermedad de Alzheimer/genética , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Animales , Diferenciación Celular , Humanos
12.
JAMA Neurol ; 71(12): 1481-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25285942

RESUMEN

IMPORTANCE: Although considerable effort has been expended developing drug candidates for Alzheimer disease, none have yet succeeded owing to the lack of efficacy or to safety concerns. One potential shortcoming of current approaches to Alzheimer disease drug discovery and development is that they rely primarily on transformed cell lines and animal models that substantially overexpress wild-type or mutant proteins. It is possible that drug development failures thus far are caused in part by the limits of these approaches, which do not accurately reveal how drug candidates will behave in naive human neuronal cells. OBJECTIVE: To analyze purified neurons derived from human induced pluripotent stem cells from patients carrying 3 different presenilin 1 (PS1) mutations and nondemented control individuals in the absence of any overexpression. We tested the efficacy of γ-secretase inhibitor and γ-secretase modulator (GSM) in neurons derived from both normal control and 3 PS1 mutations (A246E, H163R, and M146L). DESIGN, SETTING, AND PARTICIPANTS: Adult human skin biopsies were obtained from volunteers at the Alzheimer Disease Research Center, University of California, San Diego. Cell cultures were treated with γ-secretase inhibitor or GSM. Comparisons of total ß-amyloid (Aß) and Aß peptides 38, 40, and 42 in the media were made between vehicle- vs drug-treated cultures. MAIN OUTCOMES AND MEASURES: Soluble Aß levels in the media were measured by enzyme-linked immunosorbent assay. RESULTS: As predicted, mutant PS1 neurons exhibited an elevated Aß42:Aß40 ratio (P < .05) at the basal state as compared with the nondemented control neurons. Treatment with a potent non-nonsteroidal anti-inflammatory druglike GSM revealed a new biomarker signature that differs from all previous cell types and animals tested. This new signature was the same in both the mutant and control neurons and consisted of a reduction in Aß42, Aß40, and Aß38 and in the Aß42:Aß40 ratio, with no change in the total Aß levels. CONCLUSIONS AND RELEVANCE: This biomarker discrepancy is likely due to overexpression of amyloid precursor protein in the transformed cellular models. Our results suggest that biomarker signatures obtained with such models are misleading and that human neurons derived from human induced pluripotent stem cells provide a unique signature that will more accurately reflect drug response in human patients and in cerebrospinal fluid biomarker changes observed during GSM treatment.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Células Madre Pluripotentes Inducidas/citología , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Alanina/análogos & derivados , Alanina/farmacología , Péptidos beta-Amiloides/efectos de los fármacos , Antiinflamatorios/farmacología , Azepinas/farmacología , Biomarcadores/metabolismo , Heterocigoto , Humanos , Mutación/genética , Neuronas/enzimología , Fragmentos de Péptidos/efectos de los fármacos
13.
Neuron ; 83(4): 789-96, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25123310

RESUMEN

Human induced pluripotent stem cells (iPSCs) from a healthy 86-year-old male were differentiated into neural stem cells and grafted into adult immunodeficient rats after spinal cord injury. Three months after C5 lateral hemisections, iPSCs survived and differentiated into neurons and glia and extended tens of thousands of axons from the lesion site over virtually the entire length of the rat CNS. These iPSC-derived axons extended through adult white matter of the injured spinal cord, frequently penetrating gray matter and forming synapses with rat neurons. In turn, host supraspinal motor axons penetrated human iPSC grafts and formed synapses. These findings indicate that intrinsic neuronal mechanisms readily overcome the inhibitory milieu of the adult injured spinal cord to extend many axons over very long distances; these capabilities persist even in neurons reprogrammed from very aged human cells.


Asunto(s)
Axones/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/cirugía , Anciano de 80 o más Años , Animales , Axones/ultraestructura , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Vértebras Cervicales , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/ultraestructura , Masculino , Ratas
14.
Cell Rep ; 5(4): 974-85, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24239350

RESUMEN

Presenilin 1 (PS1) is the catalytic core of γ-secretase, which cleaves type 1 transmembrane proteins, including the amyloid precursor protein (APP). PS1 also has γ-secretase-independent functions, and dominant PS1 missense mutations are the most common cause of familial Alzheimer's disease (FAD). Whether PS1 FAD mutations are gain- or loss-of-function remains controversial, primarily because most studies have relied on overexpression in mouse and/or nonneuronal systems. We used isogenic euploid human induced pluripotent stem cell lines to generate and study an allelic series of PS1 mutations, including heterozygous null mutations and homozygous and heterozygous FAD PS1 mutations. Rigorous analysis of this allelic series in differentiated, purified neurons allowed us to resolve this controversy and to conclude that FAD PS1 mutations, expressed at normal levels in the appropriate cell type, impair γ-secretase activity but do not disrupt γ-secretase-independent functions of PS1. Thus, FAD PS1 mutations do not act as simple loss of PS1 function but instead dominantly gain an activity toxic to some, but not all, PS1 functions.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Neurogénesis/genética , Células Madre Pluripotentes/enzimología , Presenilina-1/genética , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/biosíntesis , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/metabolismo , Secuencia de Bases , Línea Celular , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Humanos , Mutación , Neuronas/citología , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Células Madre Pluripotentes/citología , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de ADN
15.
Chem Biol ; 18(5): 563-8, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21609837

RESUMEN

High-throughput screening (HTS) of chemical libraries is often used for the unbiased identification of compounds interacting with G protein-coupled receptors (GPCRs), the largest family of therapeutic targets. However, current HTS methods require removing GPCRs from their native environment, which modifies their pharmacodynamic properties and biases the screen toward false positive hits. Here, we developed and validated a molecular imaging (MI) agent, NIR-mbc94, which emits near infrared (NIR) light and selectively binds to endogenously expressed cannabinoid CB(2) receptors, a recognized target for treating autoimmune diseases, chronic pain and cancer. The precision and ease of this assay allows for the HTS of compounds interacting with CB(2) receptors expressed in their native environment.


Asunto(s)
Colorantes Fluorescentes/química , Norbornanos/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB2/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Agonismo Inverso de Drogas , Ensayos Analíticos de Alto Rendimiento , Ratones , Norbornanos/química , Unión Proteica , Pirazoles/química , Receptor Cannabinoide CB2/metabolismo
16.
Nat Neurosci ; 13(8): 951-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20657592

RESUMEN

The endocannabinoid 2-arachidonoylglycerol (2-AG) regulates neurotransmission and neuroinflammation by activating CB1 cannabinoid receptors on neurons and CB2 cannabinoid receptors on microglia. Enzymes that hydrolyze 2-AG, such as monoacylglycerol lipase, regulate the accumulation and efficacy of 2-AG at cannabinoid receptors. We found that the recently described serine hydrolase alpha-beta-hydrolase domain 6 (ABHD6) also controls the accumulation and efficacy of 2-AG at cannabinoid receptors. In cells from the BV-2 microglia cell line, ABHD6 knockdown reduced hydrolysis of 2-AG and increased the efficacy with which 2-AG can stimulate CB2-mediated cell migration. ABHD6 was expressed by neurons in primary culture and its inhibition led to activity-dependent accumulation of 2-AG. In adult mouse cortex, ABHD6 was located postsynaptically and its selective inhibition allowed the induction of CB1-dependent long-term depression by otherwise subthreshold stimulation. Our results indicate that ABHD6 is a rate-limiting step of 2-AG signaling and is therefore a bona fide member of the endocannabinoid signaling system.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Encéfalo/metabolismo , Glicéridos/metabolismo , Monoacilglicerol Lipasas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Transducción de Señal/fisiología , Animales , Células COS , Línea Celular , Movimiento Celular , Chlorocebus aethiops , Endocannabinoides , Potenciales Postsinápticos Excitadores/fisiología , Técnicas de Silenciamiento del Gen , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microscopía Electrónica de Transmisión , Neuronas/metabolismo , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Transfección
17.
PLoS One ; 4(12): e8271, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-20020060

RESUMEN

PK 11195 and DAA1106 bind with high-affinity to the translocator protein (TSPO, formerly known as the peripheral benzodiazepine receptor). TSPO expression in glial cells increases in response to cytokines and pathological stimuli. Accordingly, [(11)C]-PK 11195 and [(11)C]-DAA1106 are recognized molecular imaging (MI) agents capable of monitoring changes in TSPO expression occurring in vivo and in response to various neuropathologies.Here we tested the pharmacological characteristics and TSPO-monitoring potential of two novel MI agents: NIR-conPK and NIR-6T. NIR-conPK is an analogue of PK 11195 conjugated to the near-infrared (NIR) emitting fluorophore: IRDye 800CW. NIR-6T is a DAA1106 analogue also conjugated to IRDye 800CW.We found that NIR-6T competed for [(3)H]-PK 11195 binding in astrocytoma cell homogenates with nanomolar affinity, but did not exhibit specific binding in intact astrocytoma cells in culture, indicating that NIR-6T is unlikely to constitute a useful MI agent for monitoring TSPO expression in intact cells. Conversely, we found that NIR-conPK did not compete for [(3)H]-PK 11195 binding in astrocytoma cell homogenate, but exhibited specific binding in intact astrocytoma cells in culture with nanomolar affinity, suggesting that NIR-conPK binds to a protein distinct, but related to, TSPO. Accordingly, treating intact astrocytoma cells and microglia in culture with cytokines led to significant changes in the amount of NIR-conPK specific binding without corresponding change in TSPO expression. Remarkably, the cytokine-induced changes in the protein targeted by NIR-conPK in intact microglia were selective, since IFN-gamma (but not TNFalpha and TGFbeta) increased the amount of NIR-conPK specific binding in these cells.Together these results suggest that NIR-conPK binds to a protein that is related to TSPO, and expressed by astrocytomas and microglia. Our results also suggest that the expression of this protein is increased by specific cytokines, and thus allows for the monitoring of a particular subtype of microglia activation.


Asunto(s)
Astrocitoma/metabolismo , Colorantes Fluorescentes/metabolismo , Indoles/metabolismo , Microglía/metabolismo , Receptores de GABA-A/metabolismo , Animales , Extractos Celulares , Quimiocinas/metabolismo , Regulación de la Expresión Génica , Cinética , Ratones , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de GABA-A/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA